Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(3)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38339065

RESUMEN

Stroke results in neuronal cell death, which causes long-term disabilities in adults. Treatment options are limited and rely on a narrow window of opportunity. Apoptosis inhibitors demonstrate efficacy in improving neuronal cell survival in animal models of stroke. However, many inhibitors non-specifically target apoptosis pathways and high doses are needed for treatment. We explored the use of a novel caspase-3/7 inhibitor, New World Laboratories (NWL) 283, with a lower IC50 than current caspase-3/7 inhibitors. We performed in vitro and in vivo assays to determine the efficacy of NWL283 in modulating cell death in a preclinical model of stroke. In vitro and in vivo assays show that NWL283 enhances cell survival of neural precursor cells. Delivery of NWL283 following stroke enhances endogenous NPC migration and leads to increased neurogenesis in the stroke-injured cortex. Furthermore, acute NWL283 administration is neuroprotective at the stroke injury site, decreasing neuronal cell death and reducing microglia activation. Coincident with NWL283 delivery for 8 days, stroke-injured mice exhibited improved functional outcomes that persisted following cessation of the drug. Therefore, we propose that NWL283 is a promising therapeutic warranting further investigation to enhance stroke recovery.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Células-Madre Neurales , Accidente Cerebrovascular , Animales , Ratones , Supervivencia Celular , Caspasa 3 , Accidente Cerebrovascular/tratamiento farmacológico , Apoptosis , Neurogénesis/fisiología , Ratones Endogámicos C57BL , Isquemia Encefálica/tratamiento farmacológico
2.
Int J Mol Sci ; 25(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38928255

RESUMEN

Activation of neural stem cells (NSCs) correlates with improved functional outcomes in mouse models of injury. In the murine brain, NSCs have been extensively characterized and comprise (1) primitive NSCs (pNSCs) and (2) definitive NSCs (dNSCs). pNSCs are the earliest cells in the NSC lineage giving rise to dNSCs in the embryonic and adult mouse brain. pNSCs are quiescent under baseline conditions and can be activated upon injury. Herein, we asked whether human pNSCs and dNSCs can be isolated during the maturation of human cerebral organoids (COs) and activated by drugs known to regulate mouse NSC behavior. We demonstrate that self-renewing, multipotent pNSC and dNSC populations are present in human COs and express genes previously characterized in mouse NSCs. The drug NWL283, an inhibitor of apoptosis, reduced cell death in COs but did not improve NSC survival. Metformin, a drug used to treat type II diabetes that is known to promote NSC activation in mice, was found to expand human NSC pools. Together, these findings are the first to identify and characterize human pNSCs, advancing our understanding of the human NSC lineage and highlighting drugs that enhance their activity.


Asunto(s)
Células-Madre Neurales , Organoides , Humanos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Organoides/metabolismo , Organoides/citología , Organoides/efectos de los fármacos , Animales , Ratones , Diferenciación Celular , Metformina/farmacología , Células Cultivadas , Encéfalo/metabolismo , Encéfalo/citología
3.
J Neuroinflammation ; 19(1): 146, 2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35705953

RESUMEN

BACKGROUND: Neonatal stroke is a devastating insult that can lead to life-long impairments. In response to hypoxic-ischaemic injury, there is loss of neurons and glia as well as a neuroinflammatory response mediated by resident immune cells, including microglia and astrocytes, which can exacerbate damage. Administration of the antidiabetic drug metformin has been shown to improve functional outcomes in preclinical models of brain injury and the cellular basis for metformin-mediated recovery is unknown. Given metformin's demonstrated anti-inflammatory properties, we investigated its role in regulating the microglia activation and used a microglia ablation strategy to investigate the microglia-mediated outcomes in a mouse model of neonatal stroke. METHODS: Hypoxia-ischaemia (H-I) was performed on post-natal day 8. Metformin was administered for one week, starting one day after injury. Immunohistochemistry was used to examine the spatiotemporal response of microglia and astrocytes after hypoxia-ischaemia, with or without metformin treatment. To evaluate the effects of microglia depletion after hypoxia-ischaemia, we delivered Plexxikon 5622 for 1 or 2 weeks post-injury. The regional pattern of microglia and astrocyte depletion was assessed through immunohistochemistry. Motor behaviour was assessed with the righting reflex, hindlimb suspension, grip strength and cylinder tests. RESULTS: Herein, we revealed a spatiotemporally regulated response of microglia and astrocytes after hypoxia-ischaemia. Metformin treatment after hypoxia-ischaemia had no effect on microglia number and proliferation, but significantly reduced microglia activation in all regions examined, concomitant with improved behavioural outcomes in injured mice. Plexxikon 5622 treatment successfully ablated microglia, resulting in a > 90% depletion in microglia in the neonatal brain. Microglia rapidly repopulated upon treatment cessation of Plexxikon. Most interesting, microglia ablation was sufficient to reduce functional deficits after hypoxia-ischaemia, mimicking the effects of 1 week of metformin treatment post-injury. CONCLUSION: These results highlight the importance of regulating the neuroinflammatory response after neonatal stroke to promote recovery.


Asunto(s)
Hipoxia-Isquemia Encefálica , Metformina , Accidente Cerebrovascular , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Hipoxia/complicaciones , Hipoxia-Isquemia Encefálica/complicaciones , Metformina/farmacología , Metformina/uso terapéutico , Ratones , Microglía , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico
4.
Stem Cells ; 39(6): 776-786, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33529418

RESUMEN

Neural stem and progenitor cells (collectively termed neural precursor cells [NPCs]) are found along the ventricular neuraxis extending from the spinal cord to the forebrain in regionally distinct niches comprised of different cell types, architecture, and cell-cell interactions. An understanding of the factors that regulate NPC behavior is critical for developing therapeutics to repair the injured central nervous system. Herein, we demonstrate that myelin basic protein (MBP), the major cytoplasmic protein constituent of the myelin sheath in oligodendrocytes, can regulate NPC behavior. Under physiological conditions, NPCs are not in contact with intracellular MBP; however, upon injury, MBP is released into the neural parenchyma. We reveal that MBP presented in a spinal cord niche is inhibitory to NPC proliferation. This inhibitory effect is regionally distinct as spinal cord NPCs, but not forebrain-derived NPCs, are inhibited by MBP. We performed coculture and conditioned media experiments that reveal the stem cell niche is a key regulator of MBP's inhibitory actions on NPCs. The inhibition is mediated by a heat-labile protein released by spinal cord niche cells, but not forebrain niche cells. However, forebrain NPCs are also inhibited by the spinal cord derived factor as revealed following in vivo infusion of the spinal cord niche-derived conditioned media. Moreover, we show that MBP inhibits oligodendrogenesis from NPCs. Together, these findings highlight the role of MBP and the regionally distinct microenvironment in regulating NPC behavior which has important implications for stem cell-based regenerative strategies.


Asunto(s)
Diferenciación Celular/fisiología , Proteína Básica de Mielina/metabolismo , Células-Madre Neurales/metabolismo , Oligodendroglía/citología , Animales , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Vaina de Mielina/metabolismo , Médula Espinal/metabolismo
5.
Proc Natl Acad Sci U S A ; 116(30): 14823-14828, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31289234

RESUMEN

Microrobotics extends the reach of human-controlled machines to submillimeter dimensions. We introduce a microrobot that relies on optoelectronic tweezers (OET) that is straightforward to manufacture, can take nearly any desirable shape or form, and can be programmed to carry out sophisticated, multiaxis operations. One particularly useful program is a serial combination of "load," "transport," and "deliver," which can be applied to manipulate a wide range of micrometer-dimension payloads. Importantly, microrobots programmed in this manner are much gentler on fragile mammalian cells than conventional OET techniques. The microrobotic system described here was demonstrated to be useful for single-cell isolation, clonal expansion, RNA sequencing, manipulation within enclosed systems, controlling cell-cell interactions, and isolating precious microtissues from heterogeneous mixtures. We propose that the optoelectronic microrobotic system, which can be implemented using a microscope and consumer-grade optical projector, will be useful for a wide range of applications in the life sciences and beyond.


Asunto(s)
Micromanipulación/instrumentación , Robótica/instrumentación , Análisis de la Célula Individual/instrumentación , Electrónica/instrumentación , Electrónica/métodos , Humanos , Células MCF-7 , Microfluídica/instrumentación , Microfluídica/métodos , Micromanipulación/métodos , Imagen Óptica/instrumentación , Imagen Óptica/métodos , Robótica/métodos , Análisis de la Célula Individual/métodos
6.
Eur J Neurosci ; 53(4): 1334-1349, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33010080

RESUMEN

Neonatal stroke is a leading cause of long-term disability and currently available rehabilitation treatments are insufficient to promote recovery. Activating neural precursor cells (NPCs) in adult rodents, in combination with rehabilitation, can accelerate functional recovery following stroke. Here, we describe a novel method of constraint-induced movement therapy (CIMT) in a rodent model of neonatal stroke that leads to improved functional outcomes, and we asked whether the recovery was correlated with expansion of NPCs. A hypoxia/ischemia (H/I) injury was induced on postnatal day 8 (PND8) via unilateral carotid artery ligation followed by systemic hypoxia. One week and two weeks post-H/I, CIMT was administered in the form of 3 botulinum toxin (Botox) injections, which induced temporary paralysis in the unaffected limb. Functional recovery was assessed using the foot fault task. NPC proliferation was assessed using the neurosphere assay and EdU immunohistochemistry. We found that neonatal H/I injury alone expands the NPC pool by >2.5-fold relative to controls. We determined that using Botox injections as a method to provide CIMT results in significant functional motor recovery after H/I. However, CIMT does not lead to enhanced NPC activation or migration into the injured parenchyma in vivo. At the time of functional recovery, increased numbers of proliferating inflammatory cells were found within the injured motor cortex. Together, these findings suggest that NPC activation following CIMT does not account for the observed functional improvement and suggests that CIMT-mediated modification of the CNS inflammatory response may play a role in the motor recovery.


Asunto(s)
Corteza Motora , Células-Madre Neurales , Rehabilitación de Accidente Cerebrovascular , Accidente Cerebrovascular , Humanos , Recién Nacido , Recuperación de la Función
7.
Dev Biol ; 445(2): 256-270, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30472119

RESUMEN

The enteric nervous system is thought to originate solely from the neural crest. Transgenic lineage tracing revealed a novel population of clonal pancreatic duodenal homeobox-1 (Pdx1)-Cre lineage progenitor cells in the tunica muscularis of the gut that produced pancreatic descendants as well as neurons upon differentiation in vitro. Additionally, an in vivo subpopulation of endoderm lineage enteric neurons, but not glial cells, was seen especially in the proximal gut. Analysis of early transgenic embryos revealed Pdx1-Cre progeny (as well as Sox-17-Cre and Foxa2-Cre progeny) migrating from the developing pancreas and duodenum at E11.5 and contributing to the enteric nervous system. These results show that the mammalian enteric nervous system arises from both the neural crest and the endoderm. Moreover, in adult mice there are separate Wnt1-Cre neural crest stem cells and Pdx1-Cre pancreatic progenitors within the muscle layer of the gut.


Asunto(s)
Sistema Nervioso Entérico/embriología , Animales , Linaje de la Célula/genética , Duodeno/embriología , Duodeno/inervación , Duodeno/metabolismo , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Páncreas/embriología , Páncreas/inervación , Páncreas/metabolismo , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
8.
Cell Tissue Res ; 371(1): 125-141, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28776186

RESUMEN

Neural stem cells and their progeny reside in two distinct neurogenic niches within the mammalian brain: the subventricular zone and the dentate gyrus. The interplay between the neural stem cells and the niche in which they reside can have significant effects on cell kinetics and neurogenesis. A comprehensive understanding of the changes to the niche that occur through postnatal development and aging, as well as following injury, is relevant for developing therapeutics and interventions to promote neural repair. We discuss changes that occur within the neural stem and progenitor cell populations, the vasculature, extracellular matrix, microglia, and secreted proteins through aging which impact cell behavior within the neurogenic niches. We examine neural precursor cell and niche responses to injury in neonatal hypoxia-ischemia, juvenile cranial irradiation, and adult stroke. This review examines the interplay between the niche and stem cell behavior through aging and following injury as a means to understand intrinsic and extrinsic factors that regulate neurogenesis in vivo.


Asunto(s)
Envejecimiento/metabolismo , Lesiones Encefálicas/fisiopatología , Hipoxia-Isquemia Encefálica/fisiopatología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Nicho de Células Madre/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Irradiación Craneana , Humanos , Recién Nacido , Ratones , Microglía/citología , Neuronas/citología , Ratas
9.
Stem Cells ; 35(9): 2071-2082, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28733998

RESUMEN

Adult primitive neural stem cells (pNSCs) are a rare population of glial fibrillary acidic protein (GFAP)- Oct4+ cells in the mouse forebrain subependymal zone bordering the lateral ventricles that give rise to clonal neurospheres in leukemia inhibitory factor in vitro. pNSC neurospheres can be passaged to self-renew or give rise to GFAP+ NSCs that form neurospheres in epidermal growth factor and fibroblast growth factor 2, which we collectively refer to as definitive NSCs (dNSCs). Label retention experiments using doxycycline-inducible histone-2B (H2B)-green fluorescent protein (GFP) mice and several chase periods of up to 1 year quantified the adult pNSC cell cycle time as 3-5 months. We hypothesized that while pNSCs are not very proliferative at baseline, they may exist as a reserve pool of NSCs in case of injury. To test this function of pNSCs, we obtained conditional Oct4 knockout mice, Oct4fl/fl ;Sox1Cre (Oct4CKO ), which do not yield adult pNSC-derived neurospheres. When we ablated the progeny of pNSCs, namely all GFAP+ dNSCs, in these Oct4CKO mice, we found that dNSCs did not recover as they do in wild-type mice, suggesting that pNSCs are necessary for dNSC repopulation. Returning to the H2B-GFP mice, we observed that the cytosine ß-d-arabinofuranoside ablation of proliferating cells including dNSCs-induced quiescent pNSCs to proliferate and significantly dilute their H2B-GFP label. In conclusion, we demonstrate that pNSCs are the most quiescent stem cells in the adult brain reported to date and that their lineage position upstream of GFAP+ dNSCs allows them to repopulate a depleted neural lineage. Stem Cells 2017;35:2071-2082.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Ciclo Celular , Proteína Ácida Fibrilar de la Glía/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Animales , Proliferación Celular , Ratones Noqueados , Mitosis , Modelos Biológicos , Factores de Transcripción SOXB1/metabolismo , Esferoides Celulares/citología
10.
Stem Cells ; 35(2): 485-496, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27573615

RESUMEN

The adult mammalian forebrain comprises two distinct populations of neural stem cells (NSCs): rare, Oct4 positive, primitive NSCs (pNSCs) and definitive NSC (dNSC) which are more abundant and express GFAP. The pNSCs are upstream of the dNSCs in the neural stem cell lineage. Herein we show that pNSC and dNSC populations can also be isolated from the developing and adult spinal cord. Spinal cord derived pNSCs are similarly rare, Oct4 expressing cells that are responsive to leukemia inhibitory factor and dNSCs are 4-5X more abundant and express GFAP. We demonstrate that myelin basic protein (MBP) is inhibitory to both pNSC and dNSC derived colony formation. Similar to what is seen in the adult forebrain following injury, spinal cord injury results in a significant increase in the size of the dNSC and pNSC pools. Hence, both primitive and definitive neural stem cells can be isolated from along the embryonic and adult neuraxis in vivo and their behavior is regulated by MBP and injury. Stem Cells 2017;35:485-496.


Asunto(s)
Envejecimiento/metabolismo , Proteína Básica de Mielina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Médula Espinal/citología , Animales , Proliferación Celular , Separación Celular , Ensayo de Unidades Formadoras de Colonias , Factores de Transcripción Forkhead/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Ratones Mutantes , Ratones Transgénicos , Vaina de Mielina/metabolismo , Nestina/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Esferoides Celulares/citología , Traumatismos de la Médula Espinal/patología
11.
Brain Res ; 1822: 148648, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37890574

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease characterized by inflammation, death or damage of oligodendrocytes, and axonal degeneration. Current MS treatments are non-curative, associated with undesired side-effects, and expensive, highlighting the need for expanded therapeutic options for patients. There is great interest in developing interventions using drugs or therapeutics to reduce symptom onset and protect pre-existing myelin. Metformin is a well-tolerated drug used to treat Type 2 diabetes that has pleiotropic effects in the central nervous system (CNS), including reducing inflammation, enhancing oligodendrogenesis, increasing the survival/proliferation of neural stem cells (NSCs), and increasing myelination. Here, we investigated whether metformin administration could improve functional outcomes, modulate oligodendrocyte precursor cells (OPCs), and reduce inflammation in a well-established mouse model of MS- experimental autoimmune encephalomyelitis (EAE). Male and female mice received metformin treatment at the time of EAE induction ("acute") or upon presentation of disease symptoms ("delayed"). We found that acute metformin treatment improved functional outcomes, concomitant with reduced microglia numbers and decreased dysmyelination. Conversely, delayed metformin treatment did not improve functional outcomes. Our findings reveal that metformin administration can improve EAE outcomes when administered before symptom onset in both sexes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Encefalomielitis Autoinmune Experimental , Metformina , Esclerosis Múltiple , Humanos , Ratones , Femenino , Masculino , Animales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Metformina/farmacología , Inflamación/tratamiento farmacológico , Gravedad del Paciente , Ratones Endogámicos C57BL
12.
Biomedicines ; 12(3)2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38540276

RESUMEN

Stroke is the leading cause of adult disability worldwide. The majority of stroke survivors are left with devastating functional impairments for which few treatment options exist. Recently, a number of studies have used ectopic expression of transcription factors that direct neuronal cell fate with the intention of converting astrocytes to neurons in various models of brain injury and disease. While there have been reports that question whether astrocyte-to-neuron conversion occurs in vivo, here, we have asked if ectopic expression of the transcription factor Neurod1 is sufficient to promote improved functional outcomes when delivered in the subacute phase following endothelin-1-induced sensory-motor cortex stroke. We used an adeno-associated virus to deliver Neurod1 from the short GFAP promoter and demonstrated improved functional outcomes as early as 28 days post-stroke and persisting to at least 63 days post-stroke. Using Cre-based cell fate tracking, we showed that functional recovery correlated with the expression of neuronal markers in transduced cells by 28 days post-stroke. By 63 days post-stroke, the reporter-expressing cells comprised ~20% of all the neurons in the perilesional cortex and expressed markers of cortical neuron subtypes. Overall, our findings indicate that ectopic expression of Neurod1 in the stroke-injured brain is sufficient to enhance neural repair.

13.
J Magn Reson Imaging ; 37(6): 1409-18, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23712844

RESUMEN

PURPOSE: To evaluate the feasibility of using micron-sized superparamagnetic iron oxide particles (MPIOs) as an effective labeling agent for monitoring bone marrow-derived mesenchymal stromal cell (BMSC) migration in the brain using magnetic resonance imaging (MRI) in a rat model of stroke and whether the accumulation of MPIO-labeled BMSCs can be differentiated from the accumulation of free MPIO particles or hemoglobin breakdown at a site of neuronal damage. MATERIALS AND METHODS: In this study BMSCs were labeled with iron oxide and their pattern of migration following intravenous injection in a rat stroke model was monitored using a clinical MRI system followed by standard histopathology. The migration pattern was compared between intravenous injection of BMSCs alone, BMSCs labeled with MPIOs, and MPIO particles alone. RESULTS: The results demonstrated that while MRI was highly sensitive in the detection of iron oxide particle-containing cells in areas of neuronal ischemia, the true origin of cells containing iron oxide particles remains ambiguous. Therefore, detection of iron particles may not be a suitable strategy for the detection of BMSCs in the brain in a stroke model. CONCLUSION: This study suggests that the use of MPIOs as labeling agents are insufficient to conclusively determine the localization of iron within cells in regions of neuronal ischemia and hemorrhage.


Asunto(s)
Células de la Médula Ósea/patología , Rastreo Celular/métodos , Dextranos , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/patología , Accidente Cerebrovascular/patología , Animales , Encéfalo/patología , Encéfalo/cirugía , Medios de Contraste , Dextranos/química , Nanopartículas de Magnetita/química , Masculino , Tamaño de la Partícula , Ratas , Ratas Long-Evans , Coloración y Etiquetado/métodos , Accidente Cerebrovascular/cirugía
14.
Acta Pharmacol Sin ; 34(1): 78-90, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23064725

RESUMEN

Stem cell-based therapies for the treatment of stroke have received considerable attention. Two broad approaches to stem cell-based therapies have been taken: the transplantation of exogenous stem cells, and the activation of endogenous neural stem and progenitor cells (together termed neural precursors). Studies examining the transplantation of exogenous cells have demonstrated that neural stem and progenitor cells lead to the most clinically promising results. Endogenous activation of neural precursors has also been explored based on the fact that resident precursor cells have the inherent capacity to proliferate, migrate and differentiate into mature neurons in the uninjured adult brain. Studies have revealed that these neural precursor cell behaviours can be activated following stroke, whereby neural precursors will expand in number, migrate to the infarct site and differentiate into neurons. However, this innate response is insufficient to lead to functional recovery, making it necessary to enhance the activation of endogenous precursors to promote tissue repair and functional recovery. Herein we will discuss the current state of the stem cell-based approaches with a focus on endogenous repair to treat the stroke injured brain.


Asunto(s)
Encéfalo/citología , Encéfalo/patología , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Accidente Cerebrovascular/terapia , Animales , Encéfalo/irrigación sanguínea , Humanos , Neurogénesis , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/cirugía
15.
Stem Cells Transl Med ; 12(6): 415-428, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37209417

RESUMEN

Spinal cord injury (SCI) results in devastating patient outcomes with few treatment options. A promising approach to improve outcomes following SCI involves the activation of endogenous precursor populations including neural stem and progenitor cells (NSPCs) which are located in the periventricular zone (PVZ), and oligodendrocyte precursor cells (OPCs) found throughout the parenchyma. In the adult spinal cord, resident NSPCs are primarily mitotically quiescent and aneurogenic, while OPCs contribute to ongoing oligodendrogenesis into adulthood. Each of these populations is responsive to SCI, increasing their proliferation and migration to the site of injury; however, their activation is not sufficient to support functional recovery. Previous work has shown that administration of the FDA-approved drug metformin is effective at promoting endogenous brain repair following injury, and this is correlated with enhanced NSPC activation. Here, we ask whether metformin can promote functional recovery and neural repair following SCI in both males and females. Our results reveal that acute, but not delayed metformin administration improves functional outcomes following SCI in both sexes. The functional improvement is concomitant with OPC activation and oligodendrogenesis. Our data also reveal sex-dependent effects of metformin following SCI with increased activation of NSPCs in females and reduced microglia activation in males. Taken together, these findings support metformin as a viable therapeutic strategy following SCI and highlight its pleiotropic effects in the spinal cord.


Asunto(s)
Células-Madre Neurales , Traumatismos de la Médula Espinal , Masculino , Femenino , Humanos , Microglía , Traumatismos de la Médula Espinal/tratamiento farmacológico , Neuronas , Médula Espinal
16.
ACS Appl Mater Interfaces ; 15(1): 91-105, 2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36520607

RESUMEN

We exploit the electrostatic interactions between the positively charged neuroprotective peptide, pituitary adenylate cyclase-activating polypeptide (PACAP), and negatively charged poly(lactic-co-glycolic acid) (PLGA) nanoparticles to control PACAP release from the surface of nanoparticles dispersed in a hyaluronan-methylcellulose (HAMC) hydrogel composite. PACAP is a promising therapeutic for the treatment of neurological disorders, yet it has been difficult to deliver in vivo. Herein, the PACAP release rate was tuned by manipulating peptide adsorption onto the surface of blank nanoparticles by modifying either nanoparticle loading in the hydrogel or nanoparticle surface charge. This peptide-nanoparticle interaction was controlled by the pH-responsive carboxylic acid end terminal groups of PLGA. We further validated this system with the controlled release of a novel stabilized PACAP analog: Ac-[Ala15, Ala20]PACAP-propylamide, which masks its recognition to peptidases in circulation. Both wild-type and stabilized PACAP released from the vehicle increased the production of neuroprotective Interleukin-6 from cultured primary astrocytes. Using computational fluid dynamics methods, PACAP release from the composite was predicted based on experimentally derived adsorption isotherms, which exhibited similar release profiles to experimental data. This versatile adsorption-based system was used to deliver PACAP locally to the brains of stroke-injured mice over a 10 day period in vivo, highlighting its effectiveness for the controlled release of PACAP to the central nervous system.


Asunto(s)
Hidrogeles , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Ratones , Animales , Sistema de Administración de Fármacos con Nanopartículas , Preparaciones de Acción Retardada , Adsorción , Electricidad Estática
17.
Brain Res ; 1804: 148263, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36702184

RESUMEN

Endogenous neural stem cells and their progeny (together termed neural precursor cells (NPCs)) are promising candidates to facilitate neuroregeneration. Charge-balanced biphasic monopolar stimulation (BPMP) is a clinically relevant approach that can activate NPCs both in vitro and in vivo. Herein, we established a novel ex vivo stimulation system to optimize the efficacy of BPMP electric field (EF) application in activating endogenous NPCs. Using the ex vivo system, we discerned that cathodal amplitude of 200 µA resulted in the greatest NPC pool expansion and enhanced cathodal migration. Application of the same stimulation parameters in vivo resulted in the same NPC activation in the mouse brain. The design and implementation of the novel ex vivo model bridges the gap between in vitro and in vivo systems, enabling a moderate throughput stimulation system to explore and optimize EF parameters that can be applied to clinically relevant brain injury/disease models.


Asunto(s)
Células-Madre Neurales , Ratones , Animales , Células-Madre Neurales/fisiología , Neuronas , Estimulación Eléctrica/métodos
18.
Acta Biomater ; 171: 392-405, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37683963

RESUMEN

The delivery of electrical pulses to the brain via penetrating electrodes, known as brain stimulation, has been recognized as an effective clinical approach for treating neurological disorders. Resident brain neural precursor cells (NPCs) are electrosensitive cells that respond to electrical stimulation by expanding in number, migrating and differentiating which are important characteristics that support neural repair. Here, we report the design of a conductive cryogel brain stimulation electrode specifically developed for NPC activation. The cryogel electrode has a modulus switching mechanism permitting facile penetration and reducing the mechanical mismatch between brain tissue and the penetrating electrode. The cryogel demonstrated good in vivo biocompatibility and reduced the interfacial impedance to deliver the stimulating electric field with lower voltage under charge-balanced current controlled stimulation. An ex vivo assay reveals that electrical stimulation using the cryogel electrodes results in significant expansion in the size of NPC pool. Hence, the cryogel electrodes have the potential to be used for NPC activation to support endogenous neural repair. STATEMENT OF SIGNIFICANCE: The objective of this study is to develop a cryogel-based stimulation electrode as an alternative to traditional electrode materials to be used in regenerative medicine applications for enhancing neural regeneration in brain. The electrode offers benefits such as adaptive modulus for implantation, high charge storage and injection capacities, and modulus matching with brain tissue, allowing for stable delivery of electric field for long-term neuromodulation. The electrochemical properties of cryogel electrodes were characterized in living tissue with an ex vivo set-up, providing a deeper understanding of stimulation capacity in brain environments. The cryogel electrode is biocompatible and enables low voltage, current-controlled stimulation for effective activation of endogenous neural precursor cells, revealing their potential utility in neural stem cell-mediated brain repair.


Asunto(s)
Criogeles , Células-Madre Neurales , Electrodos , Neuronas/fisiología , Conductividad Eléctrica , Estimulación Eléctrica , Electrodos Implantados
19.
Stem Cells Dev ; 32(19-20): 606-621, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37551982

RESUMEN

The mature brain contains an incredible number and diversity of cells that are produced and maintained by heterogeneous pools of neural stem cells (NSCs). Two distinct types of NSCs exist in the developing and adult mouse brain: Glial Fibrillary Acidic Protein (GFAP)-negative primitive (p)NSCs and downstream GFAP-positive definitive (d)NSCs. To better understand the embryonic functions of NSCs, we performed clonal lineage tracing within neurospheres grown from either pNSCs or dNSCs to enrich for their most immediate downstream neural progenitor cells (NPCs). These clonal progenitor lineage tracing data allowed us to construct a hierarchy of progenitor subtypes downstream of pNSCs and dNSCs that were then validated using single-cell transcriptomics. Further, we identify Nexn as required for neuronal specification from neuron/astrocyte progenitor cells downstream of rare pNSCs. Combined, these data provide single-cell resolution of NPC lineages downstream of rare pNSCs that likely would be missed from population-level analyses in vivo.


Asunto(s)
Células-Madre Neurales , Ratones , Animales , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Encéfalo/metabolismo , Astrocitos/metabolismo , Diferenciación Celular/genética
20.
Nat Mater ; 10(10): 799-806, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21874004

RESUMEN

Three-dimensional (3D) protein-patterned scaffolds provide a more biomimetic environment for cell culture than traditional two-dimensional surfaces, but simultaneous 3D protein patterning has proved difficult. We developed a method to spatially control the immobilization of different growth factors in distinct volumes in 3D hydrogels, and to specifically guide differentiation of stem/progenitor cells therein. Stem-cell differentiation factors sonic hedgehog (SHH) and ciliary neurotrophic factor (CNTF) were simultaneously immobilized using orthogonal physical binding pairs, barnase-barstar and streptavidin-biotin, respectively. Barnase and streptavidin were sequentially immobilized using two-photon chemistry for subsequent concurrent complexation with fusion proteins barstar-SHH and biotin-CNTF, resulting in bioactive 3D patterned hydrogels. The technique should be broadly applicable to the patterning of a wide range of proteins.


Asunto(s)
Materiales Biomiméticos/química , Técnicas de Cultivo de Célula , Hidrogeles/síntesis química , Péptidos y Proteínas de Señalización Intercelular/química , Andamios del Tejido/química , Proteínas Bacterianas/química , Biotina/química , Factor Neurotrófico Ciliar/química , Proteínas Hedgehog/química , Proteínas Recombinantes de Fusión/química , Ribonucleasas/química , Sefarosa/química , Estreptavidina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA